Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2860 2023-11-23 14:25:24 |
2 format correct -2 word(s) 2858 2023-11-27 09:18:32 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Haraguchi, Y.; Chiang, T.; Yu, M. Application of Electrophysiology in Non-Macular Inherited Retinal Dystrophies. Encyclopedia. Available online: https://encyclopedia.pub/entry/51999 (accessed on 29 July 2024).
Haraguchi Y, Chiang T, Yu M. Application of Electrophysiology in Non-Macular Inherited Retinal Dystrophies. Encyclopedia. Available at: https://encyclopedia.pub/entry/51999. Accessed July 29, 2024.
Haraguchi, Yulia, Tsun-Kang Chiang, Minzhong Yu. "Application of Electrophysiology in Non-Macular Inherited Retinal Dystrophies" Encyclopedia, https://encyclopedia.pub/entry/51999 (accessed July 29, 2024).
Haraguchi, Y., Chiang, T., & Yu, M. (2023, November 23). Application of Electrophysiology in Non-Macular Inherited Retinal Dystrophies. In Encyclopedia. https://encyclopedia.pub/entry/51999
Haraguchi, Yulia, et al. "Application of Electrophysiology in Non-Macular Inherited Retinal Dystrophies." Encyclopedia. Web. 23 November, 2023.
Application of Electrophysiology in Non-Macular Inherited Retinal Dystrophies
Edit

Inherited retinal dystrophies encompass a diverse group of disorders affecting the structure and function of the retina, leading to progressive visual impairment and, in severe cases, blindness. Electrophysiology testing has emerged as a valuable tool in assessing and diagnosing those conditions, offering insights into the function of different parts of the visual pathway from retina to visual cortex and aiding in disease classification. The different applications and limitations of electrophysiology techniques, including multifocal electroretinogram (mfERG), full-field ERG (ffERG), electrooculogram (EOG), pattern electroretinogram (PERG), and visual evoked potential (VEP), in the diagnosis and management of these distinctive phenotypes are discussed.

electrophysiology multifocal electroretinogram full-field electroretinogram electrooculogram pattern electroretinogram visual evoked potential retina inherited retinal dystrophies retinitis pigmentosa progressive cone and cone-rod dystrophy

1. Retinitis Pigmentosa

Use of electrophysiology in the diagnostic workup for Retinitis pigmentosa (RP) can be valuable in the early detection and characterization of disease. The full-field electroretinogram (ffERG) may detect the early characteristic photoreceptor degeneration, even before symptoms are noticeable for patients, making it useful for the diagnosis of RP [1]. Full-field ERG recording enables the distinction between predominantly rod or cone system dysfunction (DA or LA protocols, respectively) and between generalized outer and inner retinal dysfunction. As seen in Figure 1, the ffERGs of RP patients characteristically include significantly decreased ERG amplitudes and increased latency compared to normal eyes in five ffERG protocols (DA 0.01, DA 10.0, OP, LA 3.0 and LA 3.0 30 Hz flicker) [2][3]. The LA 30 Hz, which objectively assesses cone system sensitivity, also correlates strongly with reduced visual acuity in RP patients [4]. These electrophysiologic findings correlate with clinical symptoms of nyctalopia and constriction of peripheral visual fields (rod photoreceptor dysfunction) and reduction in central vision (progressive cone photoreceptor dysfunction). Additionally, the location of the lesion affects the signal response to different extents. For example, the amplitudes of responses of rod, rod-cone, and cone [5] of typical RP patients with peripheral lesions gradually extending to the central retina are lower than those of pericentral RP patients with lesions at the major temporal arcade on the retina that relatively spare the far periphery [6].
Figure 1. Electroretiongram (ERG) recordings in early, intermediate and advanced stages of retinitis pigmentosa (RP). Vertical lines indicate the moment of stimulus flash. As the RP progresses, the amplitude of responses decreases, and the implicit time may increase. Cone dysfunction typically lags behind the onset of rod dysfunction. Eventually, the ERG—under both scotopic and photopic conditions—is extinguished (reused from Verbakel et al., 2009 with permission [7]).
In RP patients without visual acuity deterioration, the N1 and N1P1 mfERG components showed preserved central retinal function but were severely reduced outside zone 2 of the retina [8]. In RP, the mfERG delayed latency and decreased amplitude compared to controls has been shown to correspond with the regions of visual field losses [9]. The decreased amplitude of the central segment of mfERGs and increased latency of mfERG in the central retina have significant correlations to the loss of visual acuity [10]. Additionally, focal macular electroretinography amplitudes are reduced before the EZ thinning on OCT in the early stage of RP, which indicates that the focal macular ERG can be an earlier and reliable indicator of macular dysfunction [11]. However, mfERGs, like PERGs, may not be recommended as a primary outcome measure in patients with advanced RP and nonrecordable ffERG because the response in all tested areas can be very low [12]. On the electrooculogram (EOG), which measures the RPE potential in response to dark and light conditions, advanced RP is associated with a reduced light peak (Lp) to dark trough (Dt) ratio (i.e., Arden ratio) [13][14][15][16].
The combination of mfERG and multifocal visually evoked potential technique (mfVEP) helps determine if there is an impact of regional retinal function on the visually evoked cortical activity, as VEPs are generated primarily in the visual cortex and may be affected by abnormalities along the visual pathway [17]. As seen in Figure 2, significant positive correlations were found between the mfERG response amplitude density (RAD) and the mfVEP RAD, as well as between the mfERG implicit time and the mfVEP implicit time, indicating that some of the mfERG responses were quantitatively correlated with mfVEP response components at corresponding retinal locations [18]. The functional measures (mfERGs) in RP usually confirm the preserved central retinal function with severely reduced function outside of the central retina, which may not be reflected by ffERGs [19].
Figure 2. Examples of multifocal electroretinogram (mfERG) and multifocal visually evoked potential (mfVEP) recorded in one representative retinitis pigmentosa study eye. MfERG and mfVEP local responses were averaged in five retinal areas located at various degrees of eccentricity from the fovea: 0–2.5 (R1), 2.5–5 (R2), 5–10 (R3), 10–15 (R4) and 15–20 (R5) degrees. IT, implicit time; RAD, response amplitude density (reused from Parisi et al., 2009 with permission [18]).
Overall, electrophysiology testing may be a valuable tool in the early diagnosis and monitoring of RP, as ffERG can identify early characteristic photoreceptor degeneration, even before symptoms are noticeable for patients. This is useful in many cases of early RP and other IRDs in which the retina may appear normal despite clinical symptoms and visual field defects. Furthermore, mfERG and PERG allow an objective evaluation of residual cone function in retinitis pigmentosa patients, which can provide guidance for patient prognosis as it relates to progressive blindness and reduced vision-related quality of life [4]. MfERG, when combined with other techniques such as OCT and VEP, can provide useful information in monitoring macular function in RP [10]. Using a combination of structural imaging and electrophysiological investigations will likely provide better estimations of retinal function in RP patients.

2. Progressive Cone/Cone-Rod Dystrophy

Use of electrophysiology for cone dystrophies can be valuable in the early detection and monitoring of possible rod photoreceptor involvement. ffERG of pure cone dystrophies or early CORD shows normal rod responses with abnormal cone signals [20][21]. Cone dystrophy shows normal, preserved rod functions and severe cone dysfunction in ffERG [22]. ffERG can be particularly helpful in early stages when patients are asymptomatic with a normal fundus exam [23]. ERGs on family members with the GUCA1A mutation show reduced amplitudes in the 30 Hz flicker and photopic (cones) over scotopic (rod) response, as well as severely reduced bilateral P50 amplitudes on PERG [23][24]. Other ERG findings of cone dystrophy include implicit time delay with the 30 Hz cone flicker responses, delayed a- and b-wave photopic response, as well as decreased amplitudes of both a- and b-waves [23][25]. In CORDs, both rod and cone ERGs are abnormal with predominant cone abnormalities with delayed 30 Hz cone flicker ERG implicit time [26][27].
Overall, electrophysiology testing can be a valuable tool in the early diagnosis and monitoring of progressive cone dystrophy. ERG changes precede the decline in subjective visual functions [28], allowing for early detection. ERGs also can isolate signals of the two major photoreceptor types (rods and cones), which helps distinguish between the spectrum of diseases ranging from COD to CORD [25].

3. Cone Dystrophy with Supernormal Rod Response

The ERGs of cone dystrophy with supernormal rod ERG are pathognomonic and diagnostic of the disorder. Like other cone dystrophy disorders, the light-adapted (LA) 3.0 and LA 3.0 30 Hz ffERGs show significant reduction and delay in those patients [29]. PERG is always abnormal in the patients with supernormal rod ERG and can be undetectable in severe macular dysfunction, although some other cone and cone-rods dystrophies may have relatively preserved macular function and PERGs in early disease [30]. Several pathognomonic findings of this disorder occur in the rod system: (1) In response to a very dim white flash (0.002 phot cd s/m2), there is no detectable ERG response. (2) In response to a standard dark-adapted dim flash (DA 0.01), the ERG shows significant delay of implicit time and subnormal amplitude. (3) From 0.01 phot cd s/m2, b-wave amplitude increases rapidly with small increases in stimulus intensity. (4) In response to DA 10.0 ERG, a-wave shows a normal slope (normal phototransduction) at the beginning, followed by a broadened shape with smaller slope (a late negative component) [29][31].
Overall, electrophysiology testing is the primary tool in the diagnosis of CDSRR due to its pathognomonic and characteristic findings. To further work up this suspected diagnosis, genetic testing is necessary to confirm the mutations in either the KCNV2 or PDE6H genes.

4. Enhanced S-Cone Syndrome

Enhanced S-cone syndrome (ESCS) is a rare, slowly progressive retinal dystrophy characterized by nyctalopia from the first decade of life, reduced visual acuity, hyperopia, and no nystagmus [32][33]. ESCS is an autosomal recessive disorder related to mutations in NR2E3, which encodes a photoreceptor nuclear receptor transcription factor critical for rod photoreceptor development, leading to excess S (blue) cones at the expense of other photoreceptor subtypes [34]. The disease is characterized by an abnormal ratio of S (blue) to L/M (red/green) cone function, the absence of rods, and progressive retinal degeneration [35]. In addition to the variety of mutations in the NR2E3 gene that causes the autosomal recessive ESCS, autosomal dominant mutations in NR2E3 have been found to cause retinitis pigmentosa, although the distinct inheritance patterns and electrophysiological findings can help differentiate these conditions [36][37].
On fundus examination, there are characteristic deep nummular clumped pigmentary lesions around the vascular arcades with possible yellow streaks, as well as macular retinoschisis and cystoid maculopathy [38]. On OCT, there is a significantly thickened outer nuclear layer as well as macular abnormalities including foveomacular schisis and macular holes [39].
The abnormality of ERGs in ESCS is also pathognomonic of the disorder. ISCEV-standard recordings of ffERG in an ESCS patient are shown in Figure 3. ffERG dark-adapted 0.01, which is rod-specific, is almost always undetectable due to the absence of rods [40]. DA 3.0 ERG (mixed rod and cone response) is delayed and usually reduced [34]. The 30 Hz flicker ERG shows characteristically delayed implicit time and reduced amplitude [41]. S-cone specific and ON-/OFF ERGs show supernormal, higher amplitudes with simplified waveforms and delayed peak, which is pathognomonic of the disorder [42]. The pattern ERG, if detectable, usually shows a delayed and reduced P50 component [42][43]. MfERG shows preservation of central responses but delayed and reduced responses in peripheral rings [42].
Figure 3. ISCEV-standard rod-specific, maximal mixed rod and cone, 30 Hz flicker stimulus, and transient single-cone ERG waveforms are shown. The scales, with normal ranges for each condition, are listed in the bottom row (reused from Park et al., 2013 with permission [44]).
Overall, electrophysiological testing is an invaluable tool in the diagnosis of ESCS, which shows pathognomonic ERG findings and identifies patients for targeted specific molecular screening to finally confirm the diagnosis [32].

5. Bradyopsia

The ERG of bradyopsia shows diagnostic findings of the disorder but also requires testing of cone function under dark adaptation and understanding of impaired recovery after a bright flash for correct diagnosis, which needs a protocol in addition to the current ISCEV standard protocols [31]. DA 0.01 (rod-specific) ERG and DA 3.0 ERG are normal or mildly reduced, while LA 3.0 ERG is subnormal. The pattern ERG and standard LA 3.0 30 Hz flicker ERG are undetectable in all patients with the RGS9 mutation [45]. Additionally, there are important findings of the disorder: (1) a scotopic (rod) red flash ERG under dark adaptation evokes normal rod and dark-adapted cone function (but severely abnormal cone function under photopic conditions); (2) dim flicker stimulus under dark adaptation reveals normal ERG response at the beginning of stimulation, which attenuates to undetectable response after 2 s; (3) DA 10.0 ERG reveals normal response to the first bright flash, and the subsequent responses to further flashes are attenuated, which takes up to 2 min to recover [45][46]. Similarly, ERG in the mouse model lacking RGS9 shows a normal response in the first flash with reduction in subsequent flashes and prolonged recovery [47][48].
Overall, in the diagnosis of bradyopsia, electrophysiological testing is a valuable tool that can identify characteristic ERG findings. Furthermore, additional ERG protocols can characterize the functional deficiencies.

6. Bietti Crystalline Dystrophy

Electrophysiology testing is more useful in assessing the extent of the disease rather than as an initial diagnostic tool in Bietti crystalline dystrophy  (BCD). The ffERG shows varying degrees of rod and cone dysfunction, ranging from normal to reduced or undetectable amplitudes of scotopic and photopic a- and b-wave responses [49][50][51]. The ffERG implicit times are also prolonged in BCD, especially in scotopic ERG, which correlates with the clinical presentation of nyctalopia [52]. The progression of the disease may follow a rod-cone dystrophy pattern according to ffERG findings. Some studies show the correlation between ffERG and disease severity, in which the patients with RPE atrophy and intraretinal crystals confined to the posterior pole show normal or mildly affected ffERG [49]. The ffERG is more likely to be abnormal in the later course of the disease when the peripheral visual field is severely affected [53], although the ffERG can also remain normal in some patients in later stages of the disease, even with severe RPE and choroid atrophy [54].
In BCD, mfERG could be helpful in detecting focal regions of abnormal retinal function, especially in the phenotypes that predominantly affect the posterior pole with normal ffERG. Significant reductions in N1 and P1 amplitudes as well as delays in P1 (and to a lesser degree in N1) implicit times of mfERG can be seen in BCD patients [52][55], with relatively preserved peripheral responses [52][56]. Because a BCD patient with normal peripheral retina may have normal ffERG but abnormal mfERG [57], mfERG is more sensitive than ffERG in identifying the defective areas of the retina, which can be helpful in the evaluation and follow-up of these patients [52].
Overall, electrophysiological testing may not be critical in the initial diagnosis of BCD. However, it can be used to monitor the progression of retinal degeneration in BCD over time.

7. Late-Onset Retinal Degeneration (L-ORD)

Use of electrophysiology for L-ORD can be helpful in some cases to identify abnormalities in early disease [58][59][60], which often shows a generalized rod-cone pattern of dysfunction that appears to be more progressive at later stages of the disease [61][62]. In L-ORD patients, significant reduction of scotopic ERG is usually observed [63], while the photopic ffERG is affected in the later stage [62]. There is correlation between the decreased ffERG response to areas of drusenoid deposits and atrophy [59][61][62][64][65], because ffERG can still be elicited from the unaffected retinal areas [65].
In a study of L-ORD with ffERG, there was significant improvement in b-wave under DA 0.01 and a-wave under DA 3.0 after a prolonged dark adaptation of 16 h, suggesting an opportunity for treatment in patients with moderate to advanced L-ORD. It suggests the possibility of partial reversion of rod dysfunction [65].
In L-ORD, pattern ERG shows that there is also severe macular involvement in addition to rod-cone dysfunction on ffERG [60]. Therefore, a severe macular involvement can be detectable using PERG in L-ORD patients. MfERG shows reduced amplitudes in the areas of drusenoids, corresponding to areas of reduced sensitivity on microperimetry maps [66]. In three L-ORD patients, EOG showed a normal light peak/dark trough (Arden) ratio, with two cases demonstrating abnormal dark trough amplitude and delayed light peak [62].
Overall, electrophysiological testing may not be a critical diagnostic tool for L-ORD because ffERG can be normal in some patients, especially in early disease [67]. However, electrophysiological testing may be the objective complementary tool to monitor the progression of retinal degeneration as well as to identify partial reversion of rod dysfunction with extended dark adaptation in L-ORD.

8. Fundus Albipunctatus

Use of electrophysiology for fundus albipunctatus can be valuable and reliable in diagnosis and help differentiate patients who may also additionally have cone dystrophies. Most patients with fundus albipunctatus have a nonprogressive rod dysfunction without retinal vessel attenuation, or pigment clumping in the retina, which is similar to retinitis functata albescens. However, other patients with fundus albipunctatus may have cone dystrophy, leading to loss of central visual acuity and reduced cone ERG amplitudes [68]. Standard scotopic ffERG shows moderate to severe generalized rod system dysfunction. Scotopic dim flash ERGs can be undetectable or subnormal but can be normalized after prolonged dark adaptation. Scotopic bright flash ERG shows a reduced maximal response. The use of a red stimulus under dark adaptation and extended recordings in the dark-adapted state suggest dark-adapted cones as the probable source of the ERG signals. An improvement in the dark-adapted ERG response is considered a feature of Fundus albipunctatus because of the slow rate of regeneration of photopigment but can also be found in retinitis punctata albescens patients [69]. Photopic responses may have subnormal amplitudes [69][70][71].
Overall, electrophysiology testing, together with genetic testing, is the most reliable tool in the diagnosis of FA. Additional retinal imaging (fundus photographs, fundus autofluorescence, and OCT) can show characteristic findings and may aid with the diagnosis. Particularly in younger patients with characteristic features of fundus albipunctatus, cone and rod function can revert to normal after a prolonged period of dark adaptation.

9. Retinitis Punctata Albescens

Utilizing electrophysiology for retinitis punctata albescens can be helpful in the diagnosis of the disease. ffERG performed after standard dark adaptation shows moderate to severe generalized rod system dysfunction. The dark-adapted isolated rod ERG response is severely reduced or undetectable [72]. MfERG may show a reduced cone response in the central region of the tested area with no improvement after 20–24 h of dark adaptation [73].
Overall, electrophysiology testing can be helpful in distinguishing RPA from fundus albipunctatus, which most often shows stationary rod receptor dysfunction and recovery of cone and rod function after a prolonged period of dark adaptation that is not often seen in RPA. Genetic screening, as well as the presence of progressive loss of photoreceptor cell function, attenuated retinal vessels and pigmentary clumping, are also the important clinical manifestations of RPA in a diagnostic workup to exclude fundus albipunctatus.

References

  1. Hamel, C. Retinitis pigmentosa. Orphanet J. Rare Dis. 2006, 1, 40.
  2. Ebdali, S.; Hashemi, B.; Hashemi, H.; Jafarzadehpur, E.; Asgari, S. Time and frequency components of ERG responses in retinitis pigmentosa. Int. Ophthalmol. 2018, 38, 2435–2444.
  3. Hassan-Karimi, H.; Jafarzadehpur, E.; Blouri, B.; Hashemi, H.; Sadeghi, A.Z.; Mirzajani, A. Frequency Domain Electroretinography in Retinitis Pigmentosa versus Normal Eyes. J. Ophthalmic Vis. Res. 2012, 7, 34–38.
  4. Arsiwalla, T.A.; Cornish, E.E.; Nguyen, P.V.; Korsakova, M.; Ali, H.; Saakova, N.; Fraser, C.L.; Jamieson, R.V.; Grigg, J.R. Assessing Residual Cone Function in Retinitis Pigmentosa Patients. Transl. Vis. Sci. Technol. 2020, 9, 29.
  5. Karali, M.; Testa, F.; Brunetti-Pierri, R.; Di Iorio, V.; Pizzo, M.; Melillo, P.; Barillari, M.R.; Torella, A.; Musacchia, F.; D’Angelo, L.; et al. Clinical and Genetic Analysis of a European Cohort with Pericentral Retinitis Pigmentosa. Int. J. Mol. Sci. 2019, 21, 86.
  6. Huang, C.W.; Yang, J.J.; Yang, C.H.; Yang, C.M.; Hu, F.R.; Ho, T.C.; Chen, T.C. The structure-function correlation analysed by OCT and full field ERG in typical and pericentral subtypes of retinitis pigmentosa. Sci. Rep. 2021, 11, 16883.
  7. Verbakel, S.K.; van Huet, R.A.C.; Boon, C.J.F.; den Hollander, A.I.; Collin, R.W.J.; Klaver, C.C.W.; Hoyng, C.B.; Roepman, R.; Klevering, B.J. Non-syndromic retinitis pigmentosa. Prog. Retin. Eye Res. 2018, 66, 157–186.
  8. Todorova, M.G.; Turksever, C.; Schotzau, A.; Schorderet, D.F.; Valmaggia, C. Metabolic and functional changes in retinitis pigmentosa: Comparing retinal vessel oximetry to full-field electroretinography, electrooculogram and multifocal electroretinography. Acta Ophthalmol. 2016, 94, e231–e241.
  9. Hood, D.C.; Holopigian, K.; Greenstein, V.; Seiple, W.; Li, J.; Sutter, E.E.; Carr, R.E. Assessment of local retinal function in patients with retinitis pigmentosa using the multi-focal ERG technique. Vis. Res. 1998, 38, 163–179.
  10. Moschos, M.M.; Chatziralli, I.P.; Verriopoulos, G.; Triglianos, A.; Ladas, D.S.; Brouzas, D. Correlation between optical coherence tomography and multifocal electroretinogram findings with visual acuity in retinitis pigmentosa. Clin. Ophthalmol. 2013, 7, 2073–2078.
  11. Okado, S.; Koyanagi, Y.; Inooka, T.; Kominami, T.; Terasaki, H.; Nishiguchi, K.M.; Ueno, S. Assessments of Macular Function by Focal Macular Electroretinography and Static Perimetry in Eyes with Retinitis Pigmentosa. Retina 2022, 42, 2184–2193.
  12. Gerth, C.; Wright, T.; Heon, E.; Westall, C.A. Assessment of central retinal function in patients with advanced retinitis pigmentosa. Investig. Ophthalmol. Vis. Sci. 2007, 48, 1312–1318.
  13. Konieczka, K.; Bojinova, R.I.; Valmaggia, C.; Schorderet, D.F.; Todorova, M.G. Preserved functional and structural integrity of the papillomacular area correlates with better visual acuity in retinitis pigmentosa. Eye 2016, 30, 1310–1323.
  14. Arden, G.B.; Wolf, J.E. The electro-oculographic responses to alcohol and light in a series of patients with retinitis pigmentosa. Investig. Ophthalmol. Vis. Sci. 2000, 41, 2730–2734.
  15. Vingolo, E.M.; Livani, M.L.; Domanico, D.; Mendonca, R.H.; Rispoli, E. Optical coherence tomography and electro-oculogram abnormalities in X-linked retinitis pigmentosa. Doc. Ophthalmol. 2006, 113, 5–10.
  16. Pinckers, A.; van Aarem, A.; Brink, H. The electrooculogram in heterozygote carriers of Usher syndrome, retinitis pigmentosa, neuronal ceroid lipofuscinosis, senior syndrome and choroideremia. Ophthalmic Genet. 1994, 15, 25–30.
  17. Asanad, S.; Karanjia, R. Multifocal Electroretinogram; StatPearls Publishing: Treasure Island, FL, USA, 2023.
  18. Parisi, V.; Ziccardi, L.; Stifano, G.; Montrone, L.; Gallinaro, G.; Falsini, B. Impact of regional retinal responses on cortical visually evoked responses: Multifocal ERGs and VEPs in the retinitis pigmentosa model. Clin. Neurophysiol. 2010, 121, 380–385.
  19. Granse, L.; Ponjavic, V.; Andreasson, S. Full-field ERG, multifocal ERG and multifocal VEP in patients with retinitis pigmentosa and residual central visual fields. Acta Ophthalmol. Scand. 2004, 82, 701–706.
  20. Simunovic, M.P.; Moore, A.T. The cone dystrophies. Eye 1998, 12, 553–565.
  21. Szlyk, J.P.; Fishman, G.A.; Alexander, K.R.; Peachey, N.S.; Derlacki, D.J. Clinical subtypes of cone-rod dystrophy. Arch. Ophthalmol. 1993, 111, 781–788.
  22. Kamenarova, K.; Corton, M.; Garcia-Sandoval, B.; Fernandez-San Jose, P.; Panchev, V.; Avila-Fernandez, A.; Lopez-Molina, M.I.; Chakarova, C.; Ayuso, C.; Bhattacharya, S.S. Novel GUCA1A mutations suggesting possible mechanisms of pathogenesis in cone, cone-rod, and macular dystrophy patients. BioMed Res. Int. 2013, 2013, 517570.
  23. Hamel, C.P. Cone rod dystrophies. Orphanet J. Rare Dis. 2007, 2, 7.
  24. Downes, S.M.; Holder, G.E.; Fitzke, F.W.; Payne, A.M.; Warren, M.J.; Bhattacharya, S.S.; Bird, A.C. Autosomal dominant cone and cone-rod dystrophy with mutations in the guanylate cyclase activator 1A gene-encoding guanylate cyclase activating protein-1. Arch. Ophthalmol. 2001, 119, 96–105.
  25. Garafalo, A.V.; Sheplock, R.; Sumaroka, A.; Roman, A.J.; Cideciyan, A.V.; Jacobson, S.G. Childhood-onset genetic cone-rod photoreceptor diseases and underlying pathobiology. eBioMedicine 2021, 63, 103200.
  26. Michaelides, M.; Hardcastle, A.J.; Hunt, D.M.; Moore, A.T. Progressive cone and cone-rod dystrophies: Phenotypes and underlying molecular genetic basis. Surv. Ophthalmol. 2006, 51, 232–258.
  27. Wang, I.; Khan, N.W.; Branham, K.; Wissinger, B.; Kohl, S.; Heckenlively, J.R. Establishing baseline rod electroretinogram values in achromatopsia and cone dystrophy. Doc. Ophthalmol. 2012, 125, 229–233.
  28. Zahlava, J.; Lestak, J.; Karel, I. Optical coherence tomography in progressive cone dystrophy. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc. Czech Repub. 2014, 158, 628–634.
  29. Robson, A.G.; Webster, A.R.; Michaelides, M.; Downes, S.M.; Cowing, J.A.; Hunt, D.M.; Moore, A.T.; Holder, G.E. “Cone dystrophy with supernormal rod electroretinogram”: A comprehensive genotype/phenotype study including fundus autofluorescence and extensive electrophysiology. Retina 2010, 30, 51–62.
  30. Robson, A.G.; Michaelides, M.; Saihan, Z.; Bird, A.C.; Webster, A.R.; Moore, A.T.; Fitzke, F.W.; Holder, G.E. Functional characteristics of patients with retinal dystrophy that manifest abnormal parafoveal annuli of high density fundus autofluorescence; a review and update. Doc. Ophthalmol. 2008, 116, 79–89.
  31. Vincent, A.; Robson, A.G.; Holder, G.E. Pathognomonic (diagnostic) ERGs. A review and update. Retina 2013, 33, 5–12.
  32. Hull, S.; Arno, G.; Sergouniotis, P.I.; Tiffin, P.; Borman, A.D.; Chandra, A.; Robson, A.G.; Holder, G.E.; Webster, A.R.; Moore, A.T. Clinical and molecular characterization of enhanced S-cone syndrome in children. JAMA Ophthalmol. 2014, 132, 1341–1349.
  33. Marmor, M.F.; Jacobson, S.G.; Foerster, M.H.; Kellner, U.; Weleber, R.G. Diagnostic clinical findings of a new syndrome with night blindness, maculopathy, and enhanced S cone sensitivity. Am. J. Ophthalmol. 1990, 110, 124–134.
  34. Pachydaki, S.I.; Klaver, C.C.; Barbazetto, I.A.; Roy, M.S.; Gouras, P.; Allikmets, R.; Yannuzzi, L.A. Phenotypic features of patients with NR2E3 mutations. Arch. Ophthalmol. 2009, 127, 71–75.
  35. Milam, A.H.; Rose, L.; Cideciyan, A.V.; Barakat, M.R.; Tang, W.X.; Gupta, N.; Aleman, T.S.; Wright, A.F.; Stone, E.M.; Sheffield, V.C.; et al. The nuclear receptor NR2E3 plays a role in human retinal photoreceptor differentiation and degeneration. Proc. Natl. Acad. Sci. USA 2002, 99, 473–478.
  36. Coppieters, F.; Leroy, B.P.; Beysen, D.; Hellemans, J.; De Bosscher, K.; Haegeman, G.; Robberecht, K.; Wuyts, W.; Coucke, P.J.; De Baere, E. Recurrent mutation in the first zinc finger of the orphan nuclear receptor NR2E3 causes autosomal dominant retinitis pigmentosa. Am. J. Hum. Genet. 2007, 81, 147–157.
  37. Escher, P.; Gouras, P.; Roduit, R.; Tiab, L.; Bolay, S.; Delarive, T.; Chen, S.; Tsai, C.C.; Hayashi, M.; Zernant, J.; et al. Mutations in NR2E3 can cause dominant or recessive retinal degenerations in the same family. Hum. Mutat. 2009, 30, 342–351.
  38. Alsalamah, A.K.; Khan, A.O.; Bakar, A.A.; Schatz, P.; Nowilaty, S.R. Recognizable Patterns of Submacular Fibrosis in Enhanced S-Cone Syndrome. Ophthalmol. Retina 2021, 5, 918–927.
  39. Sustar, M.; Perovsek, D.; Cima, I.; Stirn-Kranjc, B.; Hawlina, M.; Brecelj, J. Electroretinography and optical coherence tomography reveal abnormal post-photoreceptoral activity and altered retinal lamination in patients with enhanced S-cone syndrome. Doc. Ophthalmol. 2015, 130, 165–177.
  40. Lam, B.L.; Goldberg, J.L.; Hartley, K.L.; Stone, E.M.; Liu, M. Atypical mild enhanced S-cone syndrome with novel compound heterozygosity of the NR2E3 gene. Am. J. Ophthalmol. 2007, 144, 157–159.
  41. Naik, A.; Ratra, D.; Banerjee, A.; Dalan, D.; Jandyal, S.; Rao, G.; Sen, P.; Bhende, M.; Jayaprakash, V.; Susvar, P.; et al. Enhanced S-cone syndrome: Clinical spectrum in Indian population. Indian J. Ophthalmol. 2019, 67, 523–529.
  42. Audo, I.; Michaelides, M.; Robson, A.G.; Hawlina, M.; Vaclavik, V.; Sandbach, J.M.; Neveu, M.M.; Hogg, C.R.; Hunt, D.M.; Moore, A.T.; et al. Phenotypic variation in enhanced S-cone syndrome. Investig. Ophthalmol. Vis. Sci. 2008, 49, 2082–2093.
  43. De Carvalho, E.R.; Robson, A.G.; Arno, G.; Boon, C.J.F.; Webster, A.A.; Michaelides, M. Enhanced S-Cone Syndrome: Spectrum of Clinical, Imaging, Electrophysiologic, and Genetic Findings in a Retrospective Case Series of 56 Patients. Ophthalmol. Retina 2021, 5, 195–214.
  44. Park, S.P.; Hong, I.H.; Tsang, S.H.; Lee, W.; Horowitz, J.; Yzer, S.; Allikmets, R.; Chang, S. Disruption of the human cone photoreceptor mosaic from a defect in NR2E3 transcription factor function in young adults. Graefes Arch. Clin. Exp. Ophthalmol. 2013, 251, 2299–2309.
  45. Michaelides, M.; Li, Z.; Rana, N.A.; Richardson, E.C.; Hykin, P.G.; Moore, A.T.; Holder, G.E.; Webster, A.R. Novel mutations and electrophysiologic findings in RGS9- and R9AP-associated retinal dysfunction (Bradyopsia). Ophthalmology 2010, 117, 120–127.e1.
  46. Nishiguchi, K.M.; Sandberg, M.A.; Kooijman, A.C.; Martemyanov, K.A.; Pott, J.W.; Hagstrom, S.A.; Arshavsky, V.Y.; Berson, E.L.; Dryja, T.P. Defects in RGS9 or its anchor protein R9AP in patients with slow photoreceptor deactivation. Nature 2004, 427, 75–78.
  47. Lyubarsky, A.L.; Naarendorp, F.; Zhang, X.; Wensel, T.; Simon, M.I.; Pugh, E.N., Jr. RGS9-1 is required for normal inactivation of mouse cone phototransduction. Mol. Vis. 2001, 7, 71–78.
  48. Chen, C.K.; Burns, M.E.; He, W.; Wensel, T.G.; Baylor, D.A.; Simon, M.I. Slowed recovery of rod photoresponse in mice lacking the GTPase accelerating protein RGS9-1. Nature 2000, 403, 557–560.
  49. Lee, K.Y.; Koh, A.H.; Aung, T.; Yong, V.H.; Yeung, K.; Ang, C.L.; Vithana, E.N. Characterization of Bietti crystalline dystrophy patients with CYP4V2 mutations. Investig. Ophthalmol. Vis. Sci. 2005, 46, 3812–3816.
  50. Mansour, A.M.; Uwaydat, S.H.; Chan, C.C. Long-term follow-up in Bietti crystalline dystrophy. Eur. J. Ophthalmol. 2007, 17, 680–682.
  51. Usui, T.; Tanimoto, N.; Takagi, M.; Hasegawa, S.; Abe, H. Rod and cone a-waves in three cases of Bietti crystalline chorioretinal dystrophy. Am. J. Ophthalmol. 2001, 132, 395–402.
  52. Sen, P.; Ray, R.; Ravi, P. Electrophysiological findings in Bietti’s crystalline dystrophy. Clin. Exp. Optom. 2011, 94, 302–308.
  53. Saatci, A.O.; Atas, F.; Cetin, G.O.; Kayabasi, M. Diagnostic and Management Strategies of Bietti Crystalline Dystrophy: Current Perspectives. Clin. Ophthalmol. 2023, 17, 953–967.
  54. Rossi, S.; Testa, F.; Li, A.; Iorio, V.D.; Zhang, J.; Gesualdo, C.; Corte, M.D.; Chan, C.C.; Fielding Hejtmancik, J.; Simonelli, F. An atypical form of Bietti crystalline dystrophy. Ophthalmic Genet. 2011, 32, 118–121.
  55. Garcia-Garcia, G.P.; Martinez-Rubio, M.; Moya-Moya, M.A.; Perez-Santonja, J.J.; Escribano, J. Current perspectives in Bietti crystalline dystrophy. Clin. Ophthalmol. 2019, 13, 1379–1399.
  56. Lai, T.Y.; Ng, T.K.; Tam, P.O.; Yam, G.H.; Ngai, J.W.; Chan, W.M.; Liu, D.T.; Lam, D.S.; Pang, C.P. Genotype phenotype analysis of Bietti’s crystalline dystrophy in patients with CYP4V2 mutations. Investig. Ophthalmol. Vis. Sci. 2007, 48, 5212–5220.
  57. Lockhart, C.M.; Smith, T.B.; Yang, P.; Naidu, M.; Rettie, A.E.; Nath, A.; Weleber, R.; Kelly, E.J. Longitudinal characterisation of function and structure of Bietti crystalline dystrophy: Report on a novel homozygous mutation in CYP4V2. Br. J. Ophthalmol. 2018, 102, 187–194.
  58. Borooah, S.; Collins, C.; Wright, A.; Dhillon, B. Late-onset retinal macular degeneration: Clinical insights into an inherited retinal degeneration. Br. J. Ophthalmol. 2009, 93, 284–289.
  59. Ayyagari, R.; Mandal, M.N.; Karoukis, A.J.; Chen, L.; McLaren, N.C.; Lichter, M.; Wong, D.T.; Hitchcock, P.F.; Caruso, R.C.; Moroi, S.E.; et al. Late-onset macular degeneration and long anterior lens zonules result from a CTRP5 gene mutation. Investig. Ophthalmol. Vis. Sci. 2005, 46, 3363–3371.
  60. Soumplis, V.; Sergouniotis, P.I.; Robson, A.G.; Michaelides, M.; Moore, A.T.; Holder, G.E.; Webster, A.R. Phenotypic findings in C1QTNF5 retinopathy (late-onset retinal degeneration). Acta Ophthalmol. 2013, 91, e191–e195.
  61. Milam, A.H.; Curcio, C.A.; Cideciyan, A.V.; Saxena, S.; John, S.K.; Kruth, H.S.; Malek, G.; Heckenlively, J.R.; Weleber, R.G.; Jacobson, S.G. Dominant late-onset retinal degeneration with regional variation of sub-retinal pigment epithelium deposits, retinal function, and photoreceptor degeneration. Ophthalmology 2000, 107, 2256–2266.
  62. Vincent, A.; Munier, F.L.; Vandenhoven, C.C.; Wright, T.; Westall, C.A.; Heon, E. The characterization of retinal phenotype in a family with C1QTNF5-related late-onset retinal degeneration. Retina 2012, 32, 1643–1651.
  63. Kuntz, C.A.; Jacobson, S.G.; Cideciyan, A.V.; Li, Z.Y.; Stone, E.M.; Possin, D.; Milam, A.H. Sub-retinal pigment epithelial deposits in a dominant late-onset retinal degeneration. Investig. Ophthalmol. Vis. Sci. 1996, 37, 1772–1782.
  64. De Zaeytijd, J.; Coppieters, F.; De Bruyne, M.; Van Royen, J.; Roels, D.; Six, R.; Van Cauwenbergh, C.; De Baere, E.; Leroy, B.P. Longitudinal phenotypic study of late-onset retinal degeneration due to a founder variant c.562C>A p.(Pro188Thr) in the C1QTNF5 gene. Ophthalmic Genet. 2021, 42, 521–532.
  65. Papastavrou, V.T.; Bradshaw, K.R.; Aye, K.H.; Turney, C.; Browning, A.C. Improvement of retinal function in L-ORD after prolonged dark adaptation. Can. J. Ophthalmol. 2015, 50, 112–118.
  66. Borooah, S.; Papastavrou, V.; Lando, L.; Han, J.; Lin, J.H.; Ayyagari, R.; Dhillon, B.; Browning, A.C. Reticular Pseudodrusen in Late-Onset Retinal Degeneration. Ophthalmol. Retina 2021, 5, 1043–1051.
  67. Lando, L.; Borooah, S. Late-Onset Retinal Degeneration: Clinical Perspectives. Clin. Ophthalmol. 2022, 16, 3225–3246.
  68. Nakamura, M.; Hotta, Y.; Tanikawa, A.; Terasaki, H.; Miyake, Y. A high association with cone dystrophy in Fundus albipunctatus caused by mutations of the RDH5 gene. Investig. Ophthalmol. Vis. Sci. 2000, 41, 3925–3932.
  69. Sergouniotis, P.I.; Sohn, E.H.; Li, Z.; McBain, V.A.; Wright, G.A.; Moore, A.T.; Robson, A.G.; Holder, G.E.; Webster, A.R. Phenotypic variability in RDH5 retinopathy (Fundus Albipunctatus). Ophthalmology 2011, 118, 1661–1670.
  70. Nakamura, M.; Skalet, J.; Miyake, Y. RDH5 gene mutations and electroretinogram in fundus albipunctatus with or without macular dystrophy: RDH5 mutations and ERG in fundus albipunctatus. Doc. Ophthalmol. 2003, 107, 3–11.
  71. Hajali, M.; Fishman, G.A.; Dryja, T.P.; Sweeney, M.O.; Lindeman, M. Diagnosis in a patient with fundus albipunctatus and atypical fundus changes. Doc. Ophthalmol. 2009, 118, 233–238.
  72. Fishman, G.A.; Roberts, M.F.; Derlacki, D.J.; Grimsby, J.L.; Yamamoto, H.; Sharon, D.; Nishiguchi, K.M.; Dryja, T.P. Novel mutations in the cellular retinaldehyde-binding protein gene (RLBP1) associated with retinitis punctata albescens: Evidence of interfamilial genetic heterogeneity and fundus changes in heterozygotes. Arch. Ophthalmol. 2004, 122, 70–75.
  73. Granse, L.; Abrahamson, M.; Ponjavic, V.; Andreasson, S. Electrophysiological findings in two young patients with Bothnia dystrophy and a mutation in the RLBP1 gene. Ophthalmic Genet. 2001, 22, 97–105.
More
Information
Subjects: Ophthalmology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 364
Revisions: 2 times (View History)
Update Date: 27 Nov 2023
1000/1000
Video Production Service